Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Phytother Res ; 37(9): 3898-3912, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37132081

ABSTRACT

Liver fibrosis is a key global health care burden. Sclareol, isolated from Salvia sclarea, possesses various biological activities. Its effect on liver fibrosis remains unknown. This study was proposed to evaluate the antifibrotic activity of sclareol (SCL) and explore its underlying mechanisms. Stimulated hepatic stellate cells served as an in vitro liver fibrosis model. The expression of fibrotic markers was assessed by western blot and real-time PCR. Two classical animal models, bile duct-ligated rats and carbon tetrachloride-treated mice, were utilized for the in vivo experiments. The liver function and fibrosis degree were determined by serum biochemical and histopathological analyses. VEGFR2 SUMOylation was analyzed using coimmunoprecipitation assay. Our results indicated that SCL treatment restricted the profibrotic propensity of activated HSCs. In fibrotic rodents, SCL administration alleviated hepatic injury and reduced collagen accumulation. Mechanistic studies indicated that SCL downregulated the protein level of SENP1 and enhanced VEGFR2 SUMOylation in LX-2 cells, which affected its intracellular trafficking. Blockade of the interaction between VEGFR2 and STAT3 was observed, resulting in the suppression of downstream STAT3 phosphorylation. Our findings demonstrated that SCL has therapeutic efficacy against liver fibrosis through mediating VEGFR2 SUMOylation, suggesting that SCL may be a potential candidate compound for its treatment.


Subject(s)
Liver Cirrhosis , Sumoylation , Rats , Mice , Animals , Liver Cirrhosis/drug therapy , Liver , Signal Transduction , Fibrosis , Hepatic Stellate Cells
2.
Bioorg Chem ; 127: 105971, 2022 10.
Article in English | MEDLINE | ID: mdl-35749855

ABSTRACT

Liver fibrosis is an important process in chronic liver disease and is strongly related to poor prognosis. Dehydromevalonolactone (C8) is a natural product isolated from a fungus of Fusarium sp. CPCC 401218, and its pharmacological activity has never been reported before. In this study, the potential of C8 as an anti-hepatic fibrosis agent was investigated. In human hepatic stellate cell (HSC) line LX-2, C8 suppressed the increased expression of COL1A1 and α-SMA induced by TGFß1, which indicated that C8 could repress the activation of HSCs. In bile duct ligated rats, C8 administration (100 mg/kg, i.p.) markedly attenuated liver injury, fibrosis, and inflammation, and suppressed the expression of the macrophage surface marker F4/80. In terms of mechanism, C8 treatment blocked the activation of the NLRP3 inflammasome, which was stimulated by LPS and nigericin in bone marrow-derived macrophages (BMDMs) and companied by the release of active IL-1ß. In addition, the activation of LX-2 cells induced by IL-1ß released from BMDMs was also inhibited after C8 administration, which indicated that C8 repressed HSCs activation by inhibiting the activation of NLRP3 inflammasome in macrophages. Furthermore, C8 exhibited the effects of anti-fibrosis and inhibiting the expression of NLRP3 inflammasome in non-alcoholic steatohepatitis (NASH) mice. Finally, C8 can be commendably absorbed in vivo and was safe for mice at the concentration of 1000 mg/kg (p.o.). In summary, our study reveals that C8 ameliorates HSCs activation and liver fibrosis in cholestasis rats and NASH mice by inhibiting NLRP3 inflammasome in macrophages, and C8 might be a safe and effective candidate for the treatment of liver fibrosis.


Subject(s)
Inflammasomes , Mevalonic Acid/analogs & derivatives , Non-alcoholic Fatty Liver Disease , Animals , Fibrosis , Humans , Inflammasomes/metabolism , Inflammation/metabolism , Liver/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Mevalonic Acid/analysis , Mevalonic Acid/pharmacology , Mevalonic Acid/therapeutic use , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Rats
3.
Acta Pharmacol Sin ; 42(11): 1808-1820, 2021 11.
Article in English | MEDLINE | ID: mdl-34465912

ABSTRACT

Liver fibrosis is one of the most severe pathologic consequences of chronic liver diseases, and effective therapeutic strategies are urgently needed. Proton pump inhibitors (PPIs) are H+/K+-ATPase inhibitors and currently used to treat acid-related diseases such as gastric ulcers, which have shown other therapeutic effects in addition to inhibiting acid secretion. However, few studies have focused on PPIs from the perspective of inhibiting hepatic fibrosis. In the present study, we investigated the effects of pantoprazole (PPZ), a PPI, against liver fibrosis in a bile duct ligation (BDL) rat model, human hepatic stellate cell (HSC) line LX-2 and mouse primary HSCs (pHSCs), and explored the potential mechanisms underlying the effects of PPZ in vitro and in vivo. In BDL rats, administration of PPZ (150 mg· kg-1· d-1, i.p. for 14 d) significantly attenuated liver histopathological injury, collagen accumulation, and inflammatory responses, and suppressed fibrogenesis-associated gene expression including Col1a1, Acta2, Tgfß1, and Mmp-2. In LX-2 cells and mouse pHSCs, PPZ (100-300 µM) dose-dependently suppressed the levels of fibrogenic markers. We conducted transcriptome analysis and subsequent validation in PPZ-treated LX-2 cells, and revealed that PPZ inhibited the expression of Yes-associated protein (YAP) and its downstream targets such as CTGF, ID1, survivin, CYR61, and GLI2. Using YAP overexpression and silencing, we demonstrated that PPZ downregulated hepatic fibrogenic gene expression via YAP. Furthermore, we showed that PPZ promoted the proteasome-dependent degradation and ubiquitination of YAP, thus inhibiting HSC activation. Additionally, we showed that PPZ destabilized YAP by disrupting the interaction between a deubiquitinating enzyme OTUB2 and YAP, and subsequently blocked the progression of hepatic fibrosis.


Subject(s)
Bile Ducts/drug effects , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/drug therapy , Pantoprazole/therapeutic use , Proteolysis/drug effects , YAP-Signaling Proteins/agonists , Animals , Bile Ducts/metabolism , Gene Expression Profiling , HEK293 Cells , Hepatic Stellate Cells/metabolism , Humans , Ligation , Liver Cirrhosis/metabolism , Male , Pantoprazole/pharmacology , Proton Pump Inhibitors/pharmacology , Proton Pump Inhibitors/therapeutic use , Rats , Rats, Sprague-Dawley , YAP-Signaling Proteins/metabolism
4.
Br J Pharmacol ; 177(2): 372-387, 2020 01.
Article in English | MEDLINE | ID: mdl-31621893

ABSTRACT

BACKGROUND AND PURPOSE: This study investigates the antifibrotic activities and potential mechanisms of costunolide (COS), a natural sesquiterpene compound. EXPERIMENTAL APPROACH: Rats subjected to bile duct ligation and mice challenged with CCl4 were used to study the antifibrotic effects of COS in vivo. Mouse primary hepatic stellate cells (pHSCs) and human HSC line LX-2 also served as an in vitro liver fibrosis models. The expression of fibrogenic genes and signaling proteins in the neurogenic locus notch homologue protein 3 (Notch3)-hairy/enhancer of split-1 (HES1) pathway was examined using western blot and/or real-time PCR. Notch3 degradation was analysed using immunofluorescence and coimmunoprecipitation. KEY RESULTS: In animals, COS administration attenuated hepatic histopathological injury and collagen accumulation and reduced the expression of fibrogenic genes. COS time- and dose-dependently suppressed the levels of fibrotic markers in LX-2 cells and mouse pHSCs. Mechanistic studies showed COS destabilized Notch3 and subsequently inhibited the Notch3-HES1 pathway, thus inhibiting HSC activation. Furthermore, COS blocked the WW domain-containing protein 2 (WWP2)/protein phosphatase 1G (PPM1G) interaction and enhanced the effect of WWP2 on Notch3 degradation. CONCLUSIONS AND IMPLICATIONS: COS exerted potent antifibrotic effects in vitro and in vivo by disrupting the WWP2/PPM1G complex, promoting Notch3 degradation and inhibiting the Notch3/HES1 pathway. This indicates that COS may be a potential therapeutic candidate for the treatment of liver fibrosis.


Subject(s)
Hepatic Stellate Cells/drug effects , Liver Cirrhosis, Experimental/prevention & control , Liver/drug effects , Receptor, Notch3/metabolism , Sesquiterpenes/pharmacology , Ubiquitin-Protein Ligases/metabolism , Animals , Carbon Tetrachloride , Cell Line , Common Bile Duct/surgery , Gene Expression Regulation , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Ligation , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Experimental/etiology , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Male , Mice, Inbred BALB C , Proteolysis , Rats, Sprague-Dawley , Receptor, Notch3/genetics , Signal Transduction , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Ubiquitin-Protein Ligases/genetics
5.
Chem Biodivers ; 16(4): e1800646, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30706997

ABSTRACT

A new series of (sulfonamido)propanamides (6a1-6a13, 6b1-6b15, 7c1-7c5, 6d1-6d5, 6e1-6e6) was designed and synthesized. All the synthesized compounds were characterized by NMR and mass spectrometry. The target compounds were evaluated for their in vitro cytotoxic activity against hepatocellular carcinoma (HepG2), fibrosarcoma (HT-1080), mouth epidermal carcinoma (KB), and breast adenocarcinoma (MCF-7) cell lines with the sulforhodamine B (SRB) assay, with gemcitabine and mitomycin C as positive controls. Most of these compounds exhibit a more potent cytotoxic effect than the positive control group on various cancer cell lines and the most potent compound, 6a7, shows the IC50 values of 29.78±0.516 µm, 30.70±0.61 µm, and 64.89±3.09 µm in HepG2, HT-1080, KB, and MCF-7 cell lines, respectively. Thus, these compounds with potent cytotoxic activity have potential for development as new chemotherapy agents.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Propionates/pharmacology , Sulfonamides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Propionates/chemical synthesis , Propionates/chemistry , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
6.
Acta Pharmacol Sin ; 40(7): 895-907, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30573812

ABSTRACT

The manipulation of bile acid (BA) homeostasis by blocking the ileal apical Na+-dependent bile salt transporter (ASBT/SLC10A2) may have therapeutic effects in nonalcoholic fatty liver disease. We developed a novel ASBT inhibitor, an N-(3,4-o-dichlorophenyl)-2-(3-trifluoromethoxy) benzamide derivative referred to as IMB17-15, and investigated its therapeutic effects and the molecular mechanisms underlying the effects. Syrian golden hamsters were challenged with high-fat diet (HFD) to induce NAFLD and were subsequently administered 400 mg/kg IMB17-15 by gavage daily for 21 days. Serum, liver, and fecal samples were collected for further analysis. Plasma concentration-time profiles of IMB17-15 were also constructed. The human hepatocyte cell line HL-7702 was treated with Oleic acid (OA) with or without IMB17-15. Western blotting and real-time PCR were used to study the molecular mechanisms of IMB17-15. We found that IMB17-15 inhibited ASBT and subsequently suppressed ileal farnesoid X receptor (FXR) and FXR-activated fibroblast growth factor15/19 (FGF15/19) expression, which reduced the hepatic phosphorylated extracellular regulated protein kinase (ERK) and c-Jun N-terminal kinase (JNK) levels and upregulated the cholesterol 7α-hydroxylase (CYP7A1) activity. Additionally, IMB17-15 stimulated adenosine monophosphate (AMP)-activated protein kinase (AMPKα) phosphorylation and enhanced peroxisome proliferator activated receptor α (PPARα) expression and thus promoted triglyceride (TG) oxidation and high-density lipoprotein cholesterol (HDL-c) metabolism through an ASBT-independent mechanism. In conclusion, a novel ASBT inhibitor known as IMB17-15 protected hamsters against HFD-induced NFALD by manipulating BA and lipid homeostasis. IMB17-15 also reduced lipid deposition in human hepatic cell lines, indicating that it may be useful as a therapy for NAFLD patients.


Subject(s)
Benzamides/therapeutic use , Non-alcoholic Fatty Liver Disease/prevention & control , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Sulfonamides/therapeutic use , Symporters/antagonists & inhibitors , Animals , Benzamides/pharmacokinetics , Benzamides/toxicity , Cell Line , Cytokines/metabolism , Diet, High-Fat , Female , Gene Expression Regulation/drug effects , Humans , Liver/pathology , Male , Mesocricetus , Mice , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Sulfonamides/pharmacokinetics , Sulfonamides/toxicity
7.
J Antibiot (Tokyo) ; 71(11): 965-967, 2018 11.
Article in English | MEDLINE | ID: mdl-30158646

ABSTRACT

A quinoxalinone-bohemamine hybrid compound quinohemanine (1), together with 1-methyl-2(H)-quinoxalin-2-one (2), was isolated from Streptomyces sp. CPCC 200497, a producer of quinomycins and bohemamines. Compounds 1 and 2 were purified using standard chromatographic methods, and their structures were defined through interpretation of HRMS, 1D, and 2D NMR data. Both 1 and 2 displayed moderate cytotoxicity against cancer cell line HepG2.


Subject(s)
Antineoplastic Agents/pharmacology , Pyrrolizidine Alkaloids/chemistry , Quinoxalines/chemistry , Antineoplastic Agents/chemistry , Hep G2 Cells , Humans , Molecular Structure
8.
Molecules ; 23(7)2018 07 05.
Article in English | MEDLINE | ID: mdl-29976890

ABSTRACT

A series of novel matrinic thiadiazole derivatives were designed, synthesized and evaluated for their inhibitory effect on COL1A1 promotor. The SAR indicated that: (i) the introduction of a thiadiazole on the 11-side chain was beneficial for activity; (ii) a 12-N-benzyl moiety was favorable for activity. Among them, compound 6n displayed a high activity with an inhibitory rate of 39.7% at a concentration of 40 µM. It also effectively inhibited the expression of two representative collagen proteins (COL1A1 and α-SMA) on both the mRNA and protein levels and showed a high safety profile in vivo, indicating its great promise as an anti-liver fibrosis agent. Further study indicated that it might repress hepatic fibrogenesis via the TGFß/Smad pathway. This study provided powerful information for further strategic optimization and the top compound 6n was selected for further study as an ideal liver fibrosis lead for next investigation.


Subject(s)
Actins/genetics , Alkaloids/chemistry , Collagen Type I/genetics , Quinolizines/chemistry , Thiadiazoles/administration & dosage , Thiadiazoles/chemical synthesis , Actins/pharmacology , Animals , Cell Line , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Hep G2 Cells , Humans , Liver Cirrhosis/drug therapy , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Mice , Models, Biological , Molecular Structure , Signal Transduction/drug effects , Smad Proteins/metabolism , Structure-Activity Relationship , Thiadiazoles/chemistry , Thiadiazoles/pharmacology , Transforming Growth Factor beta/metabolism , Matrines
SELECTION OF CITATIONS
SEARCH DETAIL
...